WO2010129545A2 - Particules de silicium poreux d'élution de médicament - Google Patents

Particules de silicium poreux d'élution de médicament Download PDF

Info

Publication number
WO2010129545A2
WO2010129545A2 PCT/US2010/033541 US2010033541W WO2010129545A2 WO 2010129545 A2 WO2010129545 A2 WO 2010129545A2 US 2010033541 W US2010033541 W US 2010033541W WO 2010129545 A2 WO2010129545 A2 WO 2010129545A2
Authority
WO
WIPO (PCT)
Prior art keywords
particle
agent
particles
drug
porous silicon
Prior art date
Application number
PCT/US2010/033541
Other languages
English (en)
Other versions
WO2010129545A3 (fr
Inventor
Paul Ashton
Hong Guo
Jianbing Chen
Leigh Canham
Original Assignee
Psivida Us, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Psivida Us, Inc. filed Critical Psivida Us, Inc.
Priority to CA2758607A priority Critical patent/CA2758607C/fr
Priority to EP10772695.2A priority patent/EP2427179A4/fr
Priority to AU2010246067A priority patent/AU2010246067B2/en
Priority to CN2010800172084A priority patent/CN102405042A/zh
Priority to JP2012508826A priority patent/JP5858906B2/ja
Publication of WO2010129545A2 publication Critical patent/WO2010129545A2/fr
Publication of WO2010129545A3 publication Critical patent/WO2010129545A3/fr
Priority to AU2016203393A priority patent/AU2016203393B2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0048Eye, e.g. artificial tears
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0087Galenical forms not covered by A61K9/02 - A61K9/7023
    • A61K9/0097Micromachined devices; Microelectromechanical systems [MEMS]; Devices obtained by lithographic treatment of silicon; Devices comprising chips
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents

Definitions

  • Microfabrication techniques developed by the semiconductor industry have started to receive attention for developing timed pulsatile drug release systems such as presented in Santini et al. "Microchips as Controlled Drug Delivery Devices” Angew. Chem. Int. Ed. 39, 2396-2407, 2000, which is incorporated by reference herein.
  • the drug- eluting particle of the invention comprises a biodegradable porous silicon body, a reservoir formed within the porous silicon body having at least one opening to an exterior of the body, wherein the reservoir contains a drug core comprising a therapeutic or diagnostic agent, and an agent-permeable seal disposed over the at least one opening.
  • the agent-permeable seal provides for sustained release of the agent.
  • the particle is placed in vitro or in vivo.
  • the invention further provides a method for delivering an agent, e.g., via controlled and sustained administration, of a therapeutic or diagnostic agent to a patient, comprising inserting, injecting, or implanting a sustained-release drug-eluting particle of the invention at a desired location.
  • Particles of the invention may be used in the treatment or prevention of diseases.
  • the invention also provides a method of fabricating a drug-eluting particle for releasing an agent comprising: providing a silicon wafer; depositing and patterning a material on the wafer for use as an etch mask; etching a plurality of reservoirs in the wafer; introducing pores into the silicon substrate; filling the reservoirs with a drug core comprising a therapeutic or diagnostic agent; and covering the reservoirs with a seal which is permeable to the agent.
  • FIG. 1 Top down (a) and cross sectional side view (b) of a drug-eluting particle: a. the top down view depicts the porous silicon body 3 and the agent-permeable seal 5 disposed over the reservoir; b. the cross sectional side view depicts the porous silicon body 3, a reservoir 4 containing the therapeutic or diagnostic agent 6 and an agent-permeable seal 5.
  • FIG. 1 A cross-sectional view of an alternative embodiment of the drug-eluting particle in which the reservoir 4 containing the agent 6 has two openings to the exterior of the porous silicon body 3, with an agent-permeable seal 5 disposed at each opening.
  • Figure 3 Top down representation of array of drug-eluting particles prior to partitioning into individual drug-eluting particles.
  • Figure 4 Cross-sectional views of a drug-eluting particle in which the particle comprises a porous silicon body 3, a reservoir 4 containing a therapeutic agent 6 wherein the reservoir has multiple openings 11 to an exterior of the body, and a. multiple agent- permeable seals 5 disposed over the openings; or b. an agent-permeable membrane 12 disposed over the entire surface of the particle or over portions of the particle.
  • Arrows represent the pathways of the agent 's release from the reservoir.
  • Figure 5 Wafers of silicon micro-machined to produce wells of varying diameter.
  • Figure 6. In vitro release profiles of fluocinolone acetonide (FA)-releasing particles.
  • Figure 7 The release rate per unit surface area of the reservoir.
  • Figure 8 Comparison of release rates of silicon particles with those from Iluvien style particles made from either a non-erodible (polyimide) tube or a bioerodible (poly-L-lactic acid) form.
  • the present invention provides biodegradable drug-eluting particles for sustained release of a therapeutic or diagnostic agent ("the agent").
  • the particles are designed to release the agent in a controlled fashion and then biodegrade in the body, thereby avoiding the need for surgical resection of the particles.
  • the drug- eluting particles of the invention are formed from silicon that is made biodegradable through porosification. Porous silicon is biocompatible and can be eroded in, or resorbed into, a patient without significant detrimental effect.
  • the particle comprises a porous silicon body comprising a reservoir formed within the porous silicon body having at least one opening to an exterior of the body, wherein the reservoir contains a drug core comprising a therapeutic or diagnostic agent, and an agent- permeable seal disposed over the at least one opening.
  • the seal provides for sustained release of the agent when the particle is administered to a patient.
  • a patient refers to either a human or a non-human animal.
  • porous silicon body and agent-permeable seal that remain in the body after the useful life (e.g., therapeutic utility) of the particle has ended, erode under normal biological conditions to biocompatible byproducts.
  • the particles are used in an in vitro system.
  • the drug-eluting particle comprises a porous silicon body
  • the seal provides for sustained release of the agent 6 when the particle is administered to a patient.
  • the reservoir 4 has at least two openings to an exterior of the body 3, and one or more agent-permeable seals 5 disposed over the at least two openings ( Figure 2).
  • the reservoir 4 has a plurality of openings 11 to an exterior of the body 3, and agent-permeable seals 5 disposed over the multiple openings (Figure 4a).
  • the seal is a membrane disposed over a portion of the surface, such as greater than 10%, greater than 15%, greater than 20%, greater than 30%, or greater than 40% of the surface of the porous silicon body, or the entirety of the surface of the particle.
  • a reservoir 4 has two or more openings 11 to an exterior of the body 3 and a permeable membrane 12 is disposed over the entirety of the surface of the particle ( Figure 4b).
  • a permeable seal or a permeable membrane may be disposed over the at least one opening on the surface of the particle to regulate the release of the agent.
  • the permeable seal or membrane is the only passage through which the agent is released from the particle in vivo or in vitro (e.g., prior to degradation of the particle itself).
  • the particle comprises more than one reservoir such as 2, 3, 4 or 5 reservoirs.
  • each reservoir has at least one opening to the exterior of the body, with an agent-permeable seal or membrane disposed over the at least one opening.
  • each reservoir has 2 or more openings and an agent permeable seal or membrane disposed over the at least 2 openings.
  • the drug-eluting particle of the invention is administered to a patient, e.g., by injection, for treating or preventing disease.
  • treating or treatment are intended to encompass prophylaxis and amelioration of symptoms, as well as therapy and cure.
  • preventing is art-recognized, and when used in relation to a condition such as a local reoccurrence (e.g., pain), a disease such as cancer, a syndrome complex such as heart failure or any other medical condition, is well understood in the art and includes administration of a composition which reduces the frequency of, or delays the onset of, symptoms of a medical condition in a subject relative to a subject which does not receive the composition.
  • prevention of cancer includes, for example, reducing the number of detectable cancerous growths in a population of patients receiving a prophylactic treatment relative to an untreated control population, and/or delaying the appearance of detectable cancerous growths in a treated population versus an untreated control population, e.g., by a statistically and/or clinically significant amount.
  • Prevention of an infection includes for example, reducing the number of diagnoses of the infection in a treated population versus an untreated control population, and/or delaying the onset of symptoms of the infection in a treated population versus and untreated control population.
  • Prevention of pain includes, for example, reducing the magnitude of, or alternatively delaying, pain sensations experienced by subjects in a treated population versus an untreated control population.
  • the particle releases the agent in a sustained manner, e.g., such as over a period of hours, days, weeks, months or years.
  • the particles biodegrade when the complete useful life of the particle has ended, such as days, weeks, months or years following administration.
  • the particles of the invention may possess any geometric form, such as a cube, a disc, a cylinder, a rectangular box, a pyramid, or any other regular or irregular shape.
  • the particle is formed by subdividing, e.g., by controlled partitioning, a silicon wafer, e.g., such as are used in the electronics industry.
  • the particle has a first and second side corresponding to the two planar faces of a silicon wafer.
  • one planar face of the wafer is etched to form the reservoirs in the particle prior to the controlled fracture.
  • the two sides of the particle may be disposed in any orientation, e.g., substantially parallel or skew, and may have any suitable shape, e.g., concave, convex or planar.
  • the particles have at least one dimension less than 1 mm such as a dimension from 20-600 microns such as from 100-500 microns.
  • the particles may be sufficiently small to be administered by injection through a needle such as a 21 gauge, a 22 gauge, a 23 gauge, a 24 gauge, a 25 gauge, a 26 gauge, a 27 gauge, a 28 gauge, or a 29 gauge needle.
  • the particles are administered through a 25 gauge needle.
  • the patient may be administered a single particle or multiple particles in a single administration. In certain embodiments, the patient is administered dozens or hundreds of particles.
  • the present invention provides biodegradable silicon structures and their methods of manufacture for use as drug-eluting particles.
  • Porous silicon structures unlike crystalline silicon structures, have been shown to biodegrade in vitro and in vivo producing innocuous by-products, i.e., monomeric silicic acid (Si(OH) 4 ).
  • Silicic acid which is naturally present in blood plasma at levels of less than 1 mg Si/L from the dietary intake of 20-50 mg/day, is readily removed by the kidneys.
  • the rate of dissolution in biological media varies based on the pore size in the silicon.
  • Very small pores e.g., nm- ⁇ m diameters
  • porous silicon can be coated with specific materials by existing silicon processing technology for the adsorption/desorption of liquids or gasses.
  • the porous silicon can be fabricated as thin membranes, e.g., 50 ⁇ m thick, which can be bonded to other materials, such as Pyrex or glass.
  • Biodegradation refers to the chemical breakdown of materials by a physiological environment.
  • the physiological environment under which the biodegredation occurs may be within an organism or external to an organism such as in a test tube or petri dish.
  • Biocompatibility is related to the behavior of biomaterials in various contexts. The term may refer to specific properties of a material without specifying where or how the material is used (for example, that it elicits little or no immune response in a given organism, or is able to integrate with a particular cell type or tissue), or to more empirical clinical success of a whole particle in which the material or materials feature.
  • biocompatible does not necessarily indicate that the material is biologically acceptable for all applications but merely that the material is biologically acceptable for a specific application of interest.
  • Porous silicon is commonly divided into three categories based on the size of its pores: macroporous, mesoporous and microporous.
  • Macroporous silicon has a pore diameter larger than 50 nm
  • mesoporous silicon has a pore diameter between 2 and 50 nm
  • microporous silicon has a pore diameter less than 2 nm.
  • the pore size of the porous silicon affects the rate of biodegradation of the silicon; accordingly, the pore size may be selected on the basis of the intended lifespan of the particle.
  • particles of the invention comprise mesoporous silicon, macroporous silicon, microporous silicon or a combination thereof.
  • the particles comprise mesoporous silica.
  • the porous silicon of the present particle is essentially impermeable to the passage of the therapeutic or diagnostic agent.
  • impermeable means that the layer will not allow passage of the agent at a rate sufficient to provide the desired local or systemic physiological or pharmacological effect.
  • the porous silicon body is coated with an agent-impermeable coating to prevent passage of the agent through the porous silicon. Such an agent-impermeable coating may be applied to the porous silicon body prior to the introduction of the drug core into the reservoir.
  • the agent-impermeable coating is biodegradable.
  • the porous silicon body has a porosity from 4% to 90%. In certain embodiments, the body has a porosity from 20% to 70% such as a porosity from 30% to 60% such as a porosity of about 30% or of about 35% or of about 40% or of about 45% or of about 50% or of about 55% or of about 60%.
  • Porosity as used herein, is a measure of the void spaces in a material, and is measured as a fraction, between 0 and 1, or as a percentage between 0 and 100%.
  • the particles have applicability in providing a controlled and sustained release of agents effective in obtaining a desired local or systemic physiological or pharmacological effect for treating or preventing: cancer, such as cancerous primary tumors (e.g., glioblastoma); neovascularization, including ocular neovascularization; edema, including ocular edema; inflammation, including ocular inflammation; chronic pain; arthritis; rheumatic conditions; hormonal deficiencies such as diabetes and dwarfism; or immune responses such as transplant rejection.
  • cancerous primary tumors e.g., glioblastoma
  • neovascularization including ocular neovascularization
  • edema including ocular edema
  • inflammation including ocular inflammation
  • chronic pain arthritis
  • arthritis rheumatic conditions
  • hormonal deficiencies such as diabetes and dwarfism
  • immune responses such as transplant rejection.
  • a wide variety of other disease states may also be prevented or treated using the drug delivery particle of the present invention. Such disease
  • the particles are suitable for use in treating mammalian organisms infected with HIV and AID S -related opportunistic infections such as cytomegalovirus infections, toxoplasmosis, Pneumocystis carinii, and mycobacterium avium intercellular.
  • the particles are particularly suitable for treating or preventing an ocular condition such as glaucoma, proliferative vitreoretinopathy, macular edema, including diabetic macular edema, age-related macular degeneration, diabetic retinopathy, uveitis, ocular neovascularization, and ocular infection.
  • the particles are also particularly suitable for treating mammalian organisms, both human and for veterinarian use, suffering from ocular histoplasmosis, e.g., wherein the particle is inserted or injected into the vitreous of the eye.
  • the drug core comprises a therapeutic agent effective in obtaining a desired local or systemic physiological or pharmacological effect.
  • agents such as lidocaine and related compounds and benzodiazepam and related compounds; anticancer agents such as 5-fluorouracil, adriamycin and related compounds; antiinflammatory agents such as 6-mannose phosphate; anti-fungal agents such as fluconazole and related compounds; anti-viral agents such as trisodium phosphomonoformate, trifluorothymidine, acyclovir, ganciclovir, DDI, DDC, and AZT; cell transport/mobility impending agents such as colchicine, vincristine, cytochalasin B, and related compounds; antiglaucoma drugs such as beta-blockers: timolol, betaxol, atenalol, etc; immunological response to the drug core of the particles of the present invention.
  • anesthetics and pain killing agents such as lidocaine and related compounds and benz
  • systemic administration and administered systemically mean the administration of a compound, drug or other material other than directly into the central nervous system, such that it enters the patient's system and, thus, is subject to metabolism and other like processes, for example, subcutaneous administration.
  • Local administration refers to administration at the site where the target cells are present, i.e., in a specific tissue, organ, or fluid (e.g., blood, cerebrospinal fluid, etc.).
  • agents are suitable for administration to the eye and its surrounding tissues to produce a local or a systemic physiologic or pharmacologic beneficial effect.
  • neuroprotectants such as nimodipine and related compounds; antibiotics such as tetracycline, chlortetracycline, bacitracin, neomycin, polymyxin, gramicidin, oxytetracycline, chloramphenicol, gentamycin, and erythromycin; antibacterials such as sulfonamides, sulfacetamide, sulfamethizole and, sulf ⁇ soxazole; antivirals, including idoxuridine; and other antibacterial agents such as nitrofurazone and sodium propionate; antiallergenics such as antazoline, methapyriline, chlorpheniramine, pyrilamine, and prophenpyridamine; antiinflammatories such as hydrocortisone, hydrocortisone acetate
  • particles of the invention comprise a diagnostic agent.
  • Exemplary diagnostic agents for ophthalmic use include sodium fluorescein, rose bengal, methacholine, adrenaline, ***e, and atropine.
  • the particles of the invention comprise a surgical additive.
  • Exemplary surgical additives for ophthalmic use include alpha-chymotrypsin and hyaluronidase.
  • Any pharmaceutically acceptable form of any diagnostic or therapeutic agent may be employed in the practice of the present invention, e.g., a free base or free acid or a pharmaceutically acceptable salt or ester thereof.
  • Pharmaceutically acceptable salts include sulfate, lactate, acetate, stearate, hydrochloride, tartrate, maleate, and the like.
  • drug core of the drug-eluting particles of the invention comprise from 0.01 mg to 50 mg of the agent.
  • the drug-eluting particles comprise from 0.01 mg to 40 mg of the agent, such as from 0.05 mg to 20 mg of the agent.
  • particle comprises a single reservoir which contains from 0.01 to 5 mg of the agent such as from 0.05 to 2 mg, such as from 0.1 to 1 mg of the agent.
  • the particle comprises more than one reservoir wherein each reservoir contains from 0.5 to 10 mg of the agent.
  • the drug reservoir may include additional components such as pharmaceutically acceptable carriers or an additional therapeutic agent.
  • the reservoir comprises a therapeutic agent and a biocompatible polymer, such as polyvinyl alcohol (PVA), forming a drug core.
  • PVA polyvinyl alcohol
  • Such a drug core may serve to rigidify a drug solution, thus setting an otherwise liquid drug as a solid or semi-solid within the reservoir.
  • the drug core may also serve to delay release of the therapeutic agent from the particle.
  • Pharmaceutically acceptable carriers may be added to the therapeutic agent for a variety of reasons such as ease of manufacturing, stabilizing the therapeutic agent, and altering the viscosity for loading into the particles.
  • the drug core comprises the agent, a polymer and one or more carriers. In certain embodiments, the drug core comprises one or more agents. In certain embodiments, the drug core comprises one or more polymeric agents such a cross-linked polymers or cross-linked gelatins.
  • pharmaceutically acceptable carrier refers to a pharmaceutically-acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof.
  • a pharmaceutically-acceptable material such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting any subject composition or component thereof.
  • Each carrier must be "acceptable” in the sense of being compatible with the subject composition and its components and not injurious to the patient.
  • materials which may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen- free water; isotonic saline; Ringer'
  • materials compatible with body fluids with which the particles will come in contact include, but are not limited to: ethyl vinyl acetate, polyvinyl acetate, cross-linked polyvinyl alcohol, cross-linked polyvinyl butyrate, ethylene ethylacrylate copolymer, polyethyl hexylacrylate, polyvinyl chloride, polyvinyl acetals, plasticized ethylene vinylacetate copolymer, polyvinyl alcohol, polyvinyl acetate, ethylene vinylchloride copolymer, polyvinyl esters, polyvinylbutyrate, polyvinylformal, polyamides, polymethylmethacrylate, polybutylmethacrylate, plasticized polyvinyl chloride, plasticized nylon, plasticized soft nylon, plasticized polyethylene terephthalate, natural rubber
  • the composition of the agent-permeable seal or agent- permeable membrane is selected so as to allow the above-described controlled release.
  • the agent-permeable membrane of the invention is a seal that covers one or more openings of the porous silicon body and, in addition, covers surface area beyond the opening.
  • the membrane may cover an entire side or two sides of the particle, extending beyond the opening of the reservoir.
  • the membrane covers multiple openings such as 2, 3 or 4 openings
  • the reservoir covers a percentage of the surface area of the particle including the surface of the openings such as at least about 10% of the surface, at least about 20% of the surface, at least about 30%, of the surface, at least about 40% of the surface, at least about 50 % of the surface, at least about 60% of the surface or even at least about 70 % of the surface, such as from 10-90% of the surface or 10-70 % of the surface.
  • the membrane similar to the seal, serves to control the release of the agent from the particle.
  • the preferred composition of the agent-permeable seal or membrane will vary depending on such factors as the agent, the desired rate of release, and the mode of administration.
  • the identity of the agent may also be a factor in the rate of release, since the size of the molecule, its solubility, and its polarity may play a role in determining the rate of release of the agent.
  • the agent-permeable seal or membrane is impermeable to components in a biological environment such as proteins, nucleic acids, carbohydrates, lipids, cells or cellular components.
  • the agent diffuses in the direction of lower chemical potential, i.e., toward the exterior surface of the particle. At the exterior surface of the particle, equilibrium is again established.
  • a steady state flux of the effective agent will be established in accordance with Fick's Law of Diffusion.
  • the rate of passage of the drug through the material by diffusion is generally dependent on the solubility of the drug therein, as well as on the thickness of the seal or membrane. This means that selection of appropriate materials for fabricating the seal or membrane will be dependent on the particular drug to be used.
  • Sustained drug delivery devices that utilize diffusion include those disclosed in U.S. Patent 6,375,972 which is incorporated by reference herein.
  • the rate of diffusion of the therapeutic agent through a agent-permeable seal or membrane of the present invention may be determined via diffusion cell studies carried out under sink conditions.
  • concentration of drug in the receptor compartment is essentially zero when compared to the high concentration in the donor compartment. Under these conditions, the rate of drug release is given by:
  • permeability values may be obtained from the slopes of a Q versus time plot.
  • the permeability P can be related to the diffusion coefficient D, by:
  • permeable materials suitable for use as seals or membranes are described in U.S. Pat. No. 4,014,335, which is incorporated herein by reference in its entirety. These materials include, but are not limited to: cross-linked polyvinyl alcohol, polyolefms or polyvinyl chlorides or cross-linked gelatins; regenerated, insoluble, non- erodible cellulose, acylated cellulose, esterif ⁇ ed celluloses, cellulose acetate propionate, cellulose acetate butyrate, cellulose acetate phthalate, cellulose acetate diethyl- aminoacetate; polyurethanes, polycarbonates, and microporous polymers formed by co- precipitation of a polycation and a polyanion modified insoluble collagen.
  • the reservoir seal is formed from one or more cross-linked polymers such as cross-linked polyvinyl alcohol.
  • the body of the particle may be modified with agents that enhance biocompatibility or target the particle to a specific location.
  • the surface of the silicon may be coated with a biocompatibility agent, e.g., apatite, to improve biocompatiblity.
  • a biocompatibility agent e.g., apatite
  • Other exemplary silicon modifications suitable for use with the particles disclosed herein are described in U.S. Patent No. 6,666,214 incorporated herein by reference in its entirety.
  • the body of the particle comprises a targeting moiety.
  • one or more targeting moieties are bound to the surface of the particle through covalent or non-covalent forces.
  • the targeting moiety which assists the particle in localizing to a particular target area, may be selected on the basis of the particular condition or site to be monitored.
  • the targeting moiety may further comprise any of a number of different chemical entities.
  • the targeting moiety is a small molecule. Molecules which may be suitable for use as targeting moieties in the present invention include haptens, epitopes, and dsDNA fragments and analogs and derivatives thereof.
  • Such moieties bind specifically to antibodies, fragments or analogs thereof, including mimetics (for haptens and epitopes), and zinc finger proteins (for dsDNA fragments).
  • Nutrients believed to trigger receptor-mediated endocytosis and therefore useful targeting moieties include biotin, folate, riboflavin, carnitine, inositol, lipoic acid, niacin, pantothenic acid, thiamin, pyridoxal, ascorbic acid, and the lipid soluble vitamins A, D, E and K.
  • Another exemplary type of small molecule targeting moiety includes steroidal lipids, such as cholesterol, and steroidal hormones, such as estradiol, testosterone, etc.
  • the targeting moiety may comprise a protein.
  • proteins may be selected based on known characteristics of the target site or target cells.
  • the probe can be an antibody either monoclonal or polyclonal, where a corresponding antigen is displayed at the target site.
  • the targeting moiety may comprise a protein or peptidomimetic ligand capable of binding to that receptor.
  • Protein ligands of known cell surface receptors include low density lipoproteins, transferrin, insulin, fibrinolytic enzymes, anti-HER2, platelet binding proteins such as annexins, and biological response modifiers (including interleukin, interferon, erythropoietin and colony-stimulating factor).
  • a number of monoclonal antibodies that bind to a specific type of cell have been developed, including monoclonal antibodies specific for tumor- associated antigens in humans.
  • monoclonal antibodies that may be used are anti-TAC, or other interleukin-2 receptor antibodies; 9.2.27 and NR-ML-05 to the 250 kilodalton human melanoma-associated proteoglycan; and NR-LU-10 to a pancarcinoma glycoprotein.
  • An antibody employed in the present invention may be an intact (whole) molecule, a fragment thereof, or a functional equivalent thereof. Examples of antibody fragments are F(ab') 2 , Fab', Fab, and F v fragments, which may be produced by conventional methods or by genetic or protein engineering.
  • targeting moieties include sugars (e.g., glucose, fucose, galactose, mannose) that are recognized by target-specific receptors.
  • drug-eluting particles can be modified with mannose residues (e.g., attached as C-glycosides to a free nitrogen) to yield targeted particles having higher affinity binding to tumors expressing mannose receptors (e.g., glioblastomas and gangliocytomas), and bacteria, which are also known to express mannose receptors (Bertozzi, C R and M D Bednarski Carbohydrate Research 223:243 (1992); J. Am. Chem. Soc. 114:2242,5543 (1992)), as well as potentially other infectious agents.
  • Certain cells such as malignant cells and blood cells (e.g., A, AB, B, etc.) display particular carbohydrates, for which a corresponding lectin may serve as a targeting moiety.
  • the particles are partially or completely biodegradable in a patient.
  • the particle may comprise biodegradable polymers, porous silicon and a therapeutic agent, such that after the useful life of the particle, the particle completely degrades to innocuous by-products.
  • the particle may include materials that are biodegradable and non-biodegradable. In such embodiments, the non-biodegradable component may need to be removed from the body following drug release.
  • the particles have a lifespan of hours, days, weeks, months or years.
  • lifespan refers to the time between a particle's introduction into a biological environment, e.g., in vivo or in vitro, until the particle has completely biodegraded.
  • the particle may completely degrade within hours or days such as from 4 to 24 hours, such as about 5 hours, about 10 hours, about 15 hours, about 20 hours, about 1 day, or from 2 days to 2 weeks, such as about 2 days about 5 days, about 10 days, or greater than 2 weeks, such as about 15 days, or about 20 days.
  • the particles degrade over a period of weeks or months such as about 3 weeks, about 1 month, about 2 months, about 3 months, about 6 months, about 9 months, about 1 year, about 2 years or about 3 years.
  • the invention also provides methods of fabricating a drug-eluting particle for releasing an agent comprising: providing a silicon wafer; depositing and patterning a material on the wafer for use as an etch mask; etching a plurality of reservoirs in the wafer; introducing pores into the silicon substrate; filling the reservoirs with a drug core comprising a therapeutic or diagnostic agent; and covering the reservoirs with a seal which is permeable to the agent.
  • the method further comprises a controlled partitioning of the wafer into a number of discrete particles, e.g., each comprising one reservoir.
  • Methods of the invention include methods of manufacturing porous silicon particles for sustained drug release.
  • processes may begin by modifying bulk silicon wafers, such as electronics-grade silicon, by standard methods known in the art of silicon micro fabrication and microelectromechanical systems (MEMS) in particular.
  • MEMS microelectromechanical systems
  • silicon wafers with diameters in the range of 75-350 mm in diameter and 0.05-2.0 mm in thickness may be used for producing arrays of particles.
  • the wafers are 150 mm, 200 mm or 300 mm in diameter.
  • the wafers are heavily p-type wafers with resistivity in the range of 0.1-0.001 ohm cm.
  • silicon wafers are etched on one face of the wafer to generate reservoirs as described above. This may be accomplished by patterning the wafer with a mask for creating arrays of reservoirs followed by etching by any method suitable for etching silicon, e.g., wet or dry etching.
  • Photolithography which uses light to transfer a geometric pattern from a photomask to a light-sensitive chemical (photoresist, or simply "resist") on the wafer, may be used to pattern the wafer. The exposure pattern from photolithography is engraved into the material under the photoresist with chemical treatments.
  • the process creates reservoirs in the silicon wafer which are roughly cylindrical and have diameters of about 300 ⁇ m to about 120 ⁇ m such as about 320 ⁇ m, about 300 ⁇ m, about 280 ⁇ m, about 260 ⁇ m, about 250 ⁇ m, about 240 ⁇ m, about 220 ⁇ m, about 200 ⁇ m, about 180 ⁇ m, about 160 ⁇ m, or about 140 ⁇ m.
  • the reservoirs have depths of about 500 ⁇ m to about 1 mm such as about 500 ⁇ m, about 525 ⁇ m, About 550 ⁇ m, about 575 ⁇ m, about 600 ⁇ m, about 625 ⁇ m, about 650 ⁇ m, about 675 ⁇ m, about 700 ⁇ m, about 725 ⁇ m, about 750 ⁇ m, about 775 ⁇ m, about 800 ⁇ m, about 825 ⁇ m, about 850 ⁇ m, about 875 ⁇ m, or about 900 ⁇ m.
  • the reservoirs may take the form of any geometric or amorphous shape such as a cylinder, a rectangular box, a pyramid, or an amorphous shaped reservoir.
  • the diameter for non-cylindrical reservoirs refers to the largest width measurement of a reservoir.
  • the etched silicon wafer is porosif ⁇ ed by any method in the art known for porosifying silicon.
  • silicon wafers may be porosif ⁇ ed through an anodization cell.
  • a platinum cathode and silicon wafer anode are immersed in Hydrogen Fluoride (HF) electrolyte. Corrosion of the anode is produced by running electrical current through the cell. In certain embodiments, DC current is implemented.
  • the silicon wafer may be porosif ⁇ ed through stain-etching with hydrofluoric acid, nitric acid and water. The porosity of the silicon can be increased during processing through increasing current density, decreasing HF concentration and employing a thicker silicon layer.
  • etching a plurality of reservoirs in the wafer is performed prior to, e.g., immediately prior to or a step or more prior in the process, introducing pores into the silicon body.
  • introducing pores into the silicon wafer is performed prior to, e.g., immediately prior to or a step or more prior in the process, etching a plurality of reservoirs in the wafer.
  • the surface of the etched porous silicon wafers may be modified to exhibit desired characteristics.
  • the surface may be oxidized through a controlled oxidation step such as by thermal oxidation or chemical oxidizing agents.
  • the etched porous wafer is covered with a coating that is impermeable to the agent.
  • the covering of the etched porous wafer with an agent-impermeable coating is performed prior to, e.g., immediately prior to or a step or more prior in the process, the filling of the reservoirs with the drug core.
  • the membrane is a biodegradable polymer.
  • the reservoirs of the porous silicon wafer may be filled with a drug core comprising a therapeutic or diagnostic agent by methods known in the art of micromachining.
  • Methods for filling reservoirs include, but are not limited to, methods that exploit capillary forces such as solvent techniques or ink jet printing and microinjection techniques.
  • an array of microinjectors is designed and aligned to match the reservoir array and insure massive parallel injection from each nozzle to a micromachined reservoir.
  • the filled reservoirs may then be covered with an agent-permeable seal, e.g., multiple individual seals wherein one discrete seal is disposed on each opening, a permeable seal wherein one seal covers plurality of openings, or a membrane, wherein the membrane covers a portion or the entirety of the surface of the particle.
  • the opening of the reservoir is covered with a agent-permeable membrane which covers a portion of the surface of the porous silicon body, such as greater than 10%, greater than 15%, greater than 20%, greater than 30% or greater than 40% of the surface of the porous silicon body.
  • the filled reservoirs may be covered by any method known in the art, e.g., standard techniques for depositing polymeric materials.
  • Methods for depositing thin films of material with a thickness of anywhere from a few nanometers to about 100 micrometers include, but are not limited to, spray coating, dipping, electroplating, sputter deposition, physical vapour deposition (PVD) and chemical vapour deposition (CVD).
  • the permeable seal comprises one or more polymers.
  • the reservoir array e.g., wafer
  • the reservoir array may be subdivided, e.g., through a controlled partitioning, into a number of discrete particles, e.g., each comprising one reservoir.
  • the micromachining process may be modified to include formation of one or more shallow trenches between reservoirs of the array, or otherwise scoring the surface of the array to define individual particles of the invention.
  • the controlled partitioning of the array may then be induced along the shallow trenches.
  • the reservoir array may also be cut into pieces using a high precision dicing machine. Using either of the mentioned methods or any other suitable method for subdividing the array into individual particles, it will be appreciated that the resulting individual particles may each have one or more reservoirs.
  • the reservoir array is fractured into discrete particles, each of which contains the same number of reservoirs such as a single reservoir, 2 reservoirs, 3 reservoirs, 4 reservoirs, 5 reservoirs, 6 reservoirs, 7 reservoirs, 8 reservoirs, 9 reservoirs, or 10 reservoirs.
  • the array is fractured into discrete particles in which each particle has from 1-10 reservoirs.
  • different reservoirs of a particle may include different drug cores releasing different therapeutic or diagnostic agents, or each reservoir may include the same therapeutic or diagnostic agent for diffusion out of the particle.
  • Methods of the invention include the administration of particles of the invention to a patient.
  • the invention provides a method for delivering an agent, e.g., via controlled and sustained administration, of a therapeutic or diagnostic agent to a patient.
  • agent e.g., via controlled and sustained administration
  • administering means positioning, inserting, injecting, implanting or any other means for exposing the particle to a patient.
  • the route of administration may be selected based on a variety of factors including effective amount and physical phase of the agent, type of agent and preferred site of administration.
  • the method for treating a patient, e.g., mammalian organism, to obtain a desired local or systemic physiological or pharmacological effect comprises administering the sustained- release drug delivery particle of the present invention to the patient and allowing the agent to elute from the particle and come in direct contact with cells or tissues of the patient.
  • the drug delivery particle of the present invention may be administered to a patient via any route of administration known in the art.
  • routes of administration include intraocular, oral, subcutaneous, intramuscular, intraperitoneal, intranasal, dermal, into the brain, including intracranial and intradural, into the joints, including ankles, knees, hips, shoulders, elbows, wrists, directly into tumors, and the like.
  • one or more of the particles may be administered at one time, such as 2 or more particles, dozens of particles, or hundreds of particles.
  • the drug delivery system of the present invention is particularly suitable for direct implantation into the vitreous of the eye and for application to an intraocular lens.
  • the particles may be inserted at or near the site of action such as via syringe or surgical implantation.
  • the particles of the present invention are used for localized drug delivery in treating ocular conditions, primary tumors, rheumatic and arthritic conditions, and chronic pain.
  • the particles may be implanted subcutaneously, intramuscularly, intraarterially, intrathecally, or intraperitoneally.
  • the particles of the present invention are used to provide sustained systemic levels and avoid premature metabolism.
  • such particles may be administered orally.
  • the therapeutic agent is released from the particles of the invention over the course of hours or days. In certain embodiments, the therapeutic agent is released from the particle over the course of at least about one hour, about 2 hours, about 3 hours, about 5 hours, about 10 hours, about 15 hours, about 20 hours, about 1 day, about 2 days about 5 days, about 10 days, about 15 days, or at least about 20 days. In certain embodiments, the agent is released over the course of weeks, months or years such as about 1 week, about 2 weeks, about 4 weeks, about 6 weeks, about 8 weeks, about 3 months, about 4 months, about 6 months about 9 months or about 1 year.
  • an ocular particle containing fluocinolone acetonide as the effective agent in a therapeutically effective amount to reduce or prevent ocular neovascularization may be prepared.
  • Such particles may be used to effectively combat and inhibit undesirable ocular neovascularization, edema, or inflammation when surgically implanted into the vitreous of the eye. Such particles may remain in the vitreous to biodegrade after treatment is complete.
  • the preferred amount of fluocinolone acetonide used in these particles ranges from about 0.01 mg to about 15 mg. More preferably, such particles contain from about 0.1 mg to about 6 mg of fluocinolone acetonide.
  • Preferred permeable seals in accordance with the present invention are formed of polyvinyl alcohol, which is preferably cross-linked.
  • Silicon wafers have diameters in the range of 75-350 mm in diameter, such as 150 mm, 200 mm or 300 mm in diameter. They may be heavily p-type wafers with resistively in the range 0.1-0.001 ohm/cm. Fabrication begins by deposition and photolithographic patterning of an electrically insulating material onto the most polished face of the wafer. This material serves as an etch mask and is typically silicon nitride, silicon oxide or a specific polymer.
  • a pattern appropriate for creating arrays of cylindrical reservoirs is transferred into the mask material (0.3-0.5 micron thick amorphous silicon nitride deposited by PECVD) by UV photolithography and them etching, in for example, a hydrofluoric acid based solution.
  • the patterned overlay then serves as an etch mask for subsequent etching of exposed silicon. This is carried out using wet or dry etching techniques.
  • An example of a wet etch is concentrated KOH (30-40 wt%) at 80-85 0 C.
  • dry etching techniques are Reactive Ion Etching (RIE) or Ion Beam Etching (IBE). Use of these directional etching techniques allow the realization of hundreds to thousands or reservoirs per processed wafers.
  • a 200 mm diameter wafer of 0.725 mm thickness is processed to yield an array of cylindrical reservoirs of dimensions: 250 micron internal diameter and 725 micron length.
  • the spatial arrangement and spacing of the reservoirs in the wafer is varied to provide sufficient mechanical strength for subsequent dicing of the wafer into single or multiple reservoir particles in particulate form. Porosification of the reservoir walls
  • the micromachined wafer in the form of a macroporous membrane, is planarized by impregnation of an electrical insulating and HF -resistant material into the reservoir array.
  • the planarized wafer is then subjected to extended electrochemical etching ("anodisation") in a suitable electrochemical cell, in order to convert the remaining silicon (the reservoir walls) into mesoporous form.
  • electrochemical etching anodisation
  • Mesoporous silicon is known to form when silicon is anodically biased in HF-based solutions. Electrolyte flow into the micromachined reservoir volume is prevented by their temporary filling. This forces all current to flow perpendicular to the wafer surfaces, through the remaining silicon.
  • To porosify right through a patterned wafer will typically require current densities in the range of 5-500mA/cm 2 for periods of 2-5 hours, in electrolytes that contain hydrofluoric acid and a surfactant, the optimum etch conditions depending on the pre- patterning and etched array design of the wafer.
  • the planarization agent is removed by a solvent that does not dissolve mesoporous silicon.
  • the HF-etched mesoporous membrane has a silicon hydride surface chemistry. This is converted to a silicon oxide surface through a controlled oxidation step. This may be thermal oxidation, such as 500-800 0 C in air for periods ranging from 10 minutes to 5 hours such as 700 0 C for 30 minutes. It may alternatively be via the use of another oxidizing gaseous ambient or via chemical oxidizing agent such as water, hydrogen peroxide, amines or ozone.
  • the reservoir array can be filled with drug by either conventional pharmaceutical techniques or by newer microtechniques.
  • Examples of the former include methods that exploit capillary forces such as solvent techniques or ink jet printing and microinjection techniques.
  • an array of injectors is designed and aligned to match the reservoir design and ensure massive parallel injection from one nozzle per micromachined reservoir.
  • micromachining process above can include a shallow trench etch step to define and facilitate where the wafer microcapsules are to be broken through controlled fracture into a large number of discrete particles.
  • a wafer can be sawn into microcapsules, e.g., in a standard, high precision, automated dicing machine.

Abstract

L'invention porte sur une particule d'élution de médicament biodégradable utile pour l'administration d'agents de diagnostic ou thérapeutiques. Dans certains modes de réalisation, la particule d'élution de médicament de l'invention comprend un corps de silicium poreux biodégradable, un réservoir formé à l'intérieur du corps de silicium poreux pourvu au moins d'une ouverture sur l'extérieur du corps, le réservoir contenant un agent thérapeutique ou de diagnostic et un joint perméable à l'agent disposé sur la ou les ouvertures. L'invention porte en outre sur une méthode de traitement d'un patient pour obtenir un effet physiologique ou pharmacologique local ou systémique désiré comprenant l'administration d'une particule d'administration de médicament à libération prolongée de l'invention. L'invention porte également sur des procédés de fabrication d'une particule d'élution de médicament pour la libération d'agents thérapeutiques.
PCT/US2010/033541 2009-05-04 2010-05-04 Particules de silicium poreux d'élution de médicament WO2010129545A2 (fr)

Priority Applications (6)

Application Number Priority Date Filing Date Title
CA2758607A CA2758607C (fr) 2009-05-04 2010-05-04 Particules de silicium poreux d'elution de medicament
EP10772695.2A EP2427179A4 (fr) 2009-05-04 2010-05-04 Particules de silicium poreux d'élution de médicament
AU2010246067A AU2010246067B2 (en) 2009-05-04 2010-05-04 Porous silicon drug-eluting particles
CN2010800172084A CN102405042A (zh) 2009-05-04 2010-05-04 多孔硅药物洗脱颗粒
JP2012508826A JP5858906B2 (ja) 2009-05-04 2010-05-04 多孔質シリコン薬物溶出粒子
AU2016203393A AU2016203393B2 (en) 2009-05-04 2016-05-24 Porous silicon drug-eluting particles

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US17537909P 2009-05-04 2009-05-04
US61/175,379 2009-05-04

Publications (2)

Publication Number Publication Date
WO2010129545A2 true WO2010129545A2 (fr) 2010-11-11
WO2010129545A3 WO2010129545A3 (fr) 2011-03-24

Family

ID=43030532

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2010/033541 WO2010129545A2 (fr) 2009-05-04 2010-05-04 Particules de silicium poreux d'élution de médicament

Country Status (7)

Country Link
US (3) US9023896B2 (fr)
EP (1) EP2427179A4 (fr)
JP (2) JP5858906B2 (fr)
CN (3) CN105476951A (fr)
AU (2) AU2010246067B2 (fr)
CA (1) CA2758607C (fr)
WO (1) WO2010129545A2 (fr)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012061377A1 (fr) * 2010-11-01 2012-05-10 Psivida Us, Inc. Dispositifs à base de silicium pouvant être biologiquement érodés pour administration d'agents thérapeutiques
JP2014508749A (ja) * 2011-02-11 2014-04-10 サイヴィーダ ユーエス,インコーポレイテッド 抗浮腫治療薬を使用する黄斑浮腫の治療方法
US9486459B2 (en) 2009-05-04 2016-11-08 Psivida Us, Inc. Porous silicon drug-eluting particles
US9566235B2 (en) 1998-04-17 2017-02-14 Psimedica Limited Implants for administering substances and methods of producing implants
US9603801B2 (en) 2013-03-15 2017-03-28 Psivida Us, Inc. Bioerodible silicon-based compositions for delivery of therapeutic agents
US10835495B2 (en) 2012-11-14 2020-11-17 W. R. Grace & Co.-Conn. Compositions containing a biologically active material and a non-ordered inorganic oxide material and methods of making and using the same

Families Citing this family (24)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2492167C (en) 2011-06-24 2018-12-05 Nexeon Ltd Structured particles
US8685106B2 (en) 2011-11-15 2014-04-01 Abraham Lin Method of a pharmaceutical delivery system for use within a joint replacement
US10388948B2 (en) 2012-01-30 2019-08-20 Nexeon Limited Composition of SI/C electro active material
GB2499984B (en) 2012-02-28 2014-08-06 Nexeon Ltd Composite particles comprising a removable filler
EP2841528B1 (fr) * 2012-04-26 2020-01-08 King Abdullah University Of Science And Technology Silicium poreux amorphe photoluminescent colloïdal, procédés de fabrication d'un silicium poreux amorphe photoluminescent colloïdal, et procédés d'utilisation d'un silicium poreux amorphe photoluminescent colloïdal
US9878137B2 (en) 2012-05-30 2018-01-30 The Regents Of The University Of California Bioactive agent delivery devices and methods of making and using the same
GB2502625B (en) 2012-06-06 2015-07-29 Nexeon Ltd Method of forming silicon
EP2879761A4 (fr) * 2012-08-06 2016-03-02 Baylor College Medicine Dispositif d'administration de produit thérapeutique et ses procédés de fabrication
GB2507535B (en) 2012-11-02 2015-07-15 Nexeon Ltd Multilayer electrode
EP2894186A1 (fr) 2014-01-14 2015-07-15 Université de Strasbourg Matériau organométaloxide poreux désintégrable
KR101567203B1 (ko) 2014-04-09 2015-11-09 (주)오렌지파워 이차 전지용 음극 활물질 및 이의 방법
KR101604352B1 (ko) 2014-04-22 2016-03-18 (주)오렌지파워 음극 활물질 및 이를 포함하는 리튬 이차 전지
WO2016044366A1 (fr) * 2014-09-16 2016-03-24 Psivida Us, Inc. Compositions et méthodes pour la libération contrôlée d'agents
GB2533161C (en) 2014-12-12 2019-07-24 Nexeon Ltd Electrodes for metal-ion batteries
JP6785551B2 (ja) * 2015-04-03 2020-11-18 三菱製紙株式会社 エッチング方法
WO2017083779A1 (fr) 2015-11-12 2017-05-18 Graybug Vision, Inc. Aggrégation de microparticules pour un usage thérapeutique
EP3442540A4 (fr) * 2016-04-14 2019-12-18 Spinnaker Biosciences Inc. Matériaux de silicium poreux comprenant un silicate métallique pour l'administration d'agents thérapeutiques
EP3469644B1 (fr) 2016-06-14 2022-11-09 Nexeon Limited Électrodes pour des batteries à ions métalliques
WO2018162676A1 (fr) 2017-03-08 2018-09-13 Université De Strasbourg Nanoparticules d'organosilice ou d'oxyde organométallique poreuses désintégrables et leurs utilisations comme véhicule pour l'administration contrôlée d'arnsi
MX2019011242A (es) 2017-03-23 2020-01-21 Graybug Vision Inc Farmacos y composiciones para el tratamiento de trastornos oculares.
CN111201040A (zh) 2017-05-10 2020-05-26 灰色视觉公司 用于医学疗法的缓释微粒及其悬浮液
CN110913869A (zh) 2017-07-27 2020-03-24 国立大学法人大阪大学 药剂及其制造方法
WO2019246130A1 (fr) * 2018-06-19 2019-12-26 Cella Therapeutics, Llc Systèmes d'administration de médicament à libération prolongée comprenant un agent abaissant la pression intraoculaire, un composé de cnp, un composé de npr-b, un agoniste de tie-2, ou un agent neurotrophique destinés à être utilisés pour traiter le glaucome ou l'hypertension oculaire
WO2021062079A1 (fr) 2019-09-25 2021-04-01 Carbon, Inc. Procédés de revêtement de particules pour des produits fabriqués de manière additive

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009009563A2 (fr) 2007-07-10 2009-01-15 The Regents Of The University Of California Matériaux et méthodes d'administration de compositions dans des tissus sélectionnés

Family Cites Families (86)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3832458A (en) 1971-12-06 1974-08-27 River C Foundation Hydrophilic silicone composition and method
US4036979A (en) 1974-01-25 1977-07-19 American Cyanamid Company Compositions containing 4,5,6,7-tetrahydrobenz[b]thien-4-yl-ureas or derivatives and methods of enhancing growth rate
US3919723A (en) 1974-05-20 1975-11-18 Friedrichsfeld Gmbh Bone shaft or bone joint prosthesis and process
US3919060A (en) 1974-06-14 1975-11-11 Ibm Method of fabricating semiconductor device embodying dielectric isolation
DE3214667C2 (de) 1982-04-21 1985-07-18 Akzo Gmbh, 5600 Wuppertal Zusammengesetzter Körper für die Langzeitabgabe von Wirkstoffen
JPS59101145A (ja) 1982-11-30 1984-06-11 日本特殊陶業株式会社 薬液含浸多孔質セラミツクス
JPS59131346A (ja) 1983-01-17 1984-07-28 日本特殊陶業株式会社 薬液含浸多孔質セラミツクスの製造法
DE3423667A1 (de) 1984-05-08 1985-11-28 Rüdiger Dipl.-Ing. 5204 Lohmar Scheunemann Implantat fuer knochen- und zahnwurzelersatz mit spezieller uebergangsstruktur zwischen knochen und implantatkern und knochenseitig ganz oder teilweise resorbierbarer matrix
GB8422876D0 (en) 1984-09-11 1984-10-17 Secr Defence Silicon implant devices
US4608048A (en) 1984-12-06 1986-08-26 Alza Corporation Dispensing device with drug delivery patterns
US5057082A (en) 1988-11-04 1991-10-15 Plastic Injectors, Inc. Trocar assembly
DK0463061T3 (da) 1989-03-17 1993-07-12 Pitman Moore Inc Controlled-release tilførselsindretning til afgivelse af makromolekylære proteiner
US5167625A (en) 1990-10-09 1992-12-01 Sarcos Group Multiple vesicle implantable drug delivery system
US5366454A (en) 1993-03-17 1994-11-22 La Corporation De L'ecole Polytechnique Implantable medication dispensing device
US5370630A (en) 1993-11-12 1994-12-06 Smidebush; Michael J. Device for injection of fluidic materials into body tissue
US5558071A (en) 1994-03-07 1996-09-24 Combustion Electromagnetics, Inc. Ignition system power converter and controller
US5591453A (en) 1994-07-27 1997-01-07 The Trustees Of The University Of Pennsylvania Incorporation of biologically active molecules into bioactive glasses
US5817327A (en) 1994-07-27 1998-10-06 The Trustees Of The University Of Pennsylvania Incorporation of biologically active molecules into bioactive glasses
US5665114A (en) 1994-08-12 1997-09-09 Meadox Medicals, Inc. Tubular expanded polytetrafluoroethylene implantable prostheses
US5792099A (en) 1995-02-14 1998-08-11 Decamp; Dennis Syringe and cannula for insertion of viscoelastic material into an eye and method of using same
MX9707585A (es) 1995-04-03 1997-12-31 Abbott Lab Mezclas homogeneas de farmacos y aditivos de fusion a baja temperatura para liberacion controlada.
US6096344A (en) 1995-07-28 2000-08-01 Advanced Polymer Systems, Inc. Bioerodible porous compositions
GB9611437D0 (en) 1995-08-03 1996-08-07 Secr Defence Biomaterial
US5665428A (en) 1995-10-25 1997-09-09 Macromed, Inc. Preparation of peptide containing biodegradable microspheres by melt process
DE19608423A1 (de) 1996-03-05 1997-09-11 Merck Patent Gmbh Implantate mit phasenweiser Arzneistoffabgabe
US7070590B1 (en) * 1996-07-02 2006-07-04 Massachusetts Institute Of Technology Microchip drug delivery devices
US5797898A (en) 1996-07-02 1998-08-25 Massachusetts Institute Of Technology Microchip drug delivery devices
US5922299A (en) 1996-11-26 1999-07-13 Battelle Memorial Institute Mesoporous-silica films, fibers, and powders by evaporation
JP3217286B2 (ja) 1996-12-19 2001-10-09 科学技術振興事業団 Dna固定化複合体
US6696258B1 (en) 1998-01-20 2004-02-24 Drexel University Mesoporous materials and methods of making the same
AU2460099A (en) 1998-01-20 1999-08-02 Drexel University Mesoporous materials and methods of making the same
US6060036A (en) 1998-02-09 2000-05-09 Implant Sciences Corporation Radioactive seed implants
GB9808052D0 (en) 1998-04-17 1998-06-17 Secr Defence Implants for administering substances and methods of producing implants
GB9815819D0 (en) 1998-07-22 1998-09-16 Secr Defence Transferring materials into cells and a microneedle array
US5973341A (en) 1998-12-14 1999-10-26 Philips Electronics North America Corporation Lateral thin-film silicon-on-insulator (SOI) JFET device
ATE306250T1 (de) 1998-12-23 2005-10-15 Alza Corp Dosierungsformen, die poröse partikel enthalten
AU7881000A (en) 1999-08-16 2001-03-13 Henceforth Hibernia, Inc. Therapeutic and prophylactic compositions including catalytic biomimetic solids and methods to prepare and use them
GB9924334D0 (en) 1999-10-15 1999-12-15 Secr Defence Pharmaceutical products and methods of fabrication therefor
ES2182731T1 (es) 1999-10-18 2003-03-16 Ferx Inc Vehiculo magnetico para dirigir un agente biologicamente activo a un sitio especifico.
SE9903958D0 (sv) 1999-11-02 1999-11-02 Boerje Sellergren Porous materials for selective binding or transport of molecular guests
US6521284B1 (en) 1999-11-03 2003-02-18 Scimed Life Systems, Inc. Process for impregnating a porous material with a cross-linkable composition
AUPQ573300A0 (en) 2000-02-21 2000-03-16 Australian Nuclear Science & Technology Organisation Controlled release ceramic particles, compositions thereof, processes of preparation and methods of use
DE60124387T2 (de) 2000-02-28 2007-10-11 Pfizer Enterprises Sarl. Synergistische pharmazeutische Zusammensetzungen zur Behandlung von kolorektalem Krebs
IL134830A0 (en) 2000-03-01 2001-05-20 Chay 13 Medical Res Group N V Peptides and immunostimulatory and anti-bacterial pharmaceutical compositions containing them
KR20030009389A (ko) 2000-03-14 2003-01-29 브루카드 괴케 유문동-유문-십이지장 운동성에 대한 글루카곤 유사펩티드-1 (7-36)의 효과
GB0008494D0 (en) 2000-04-07 2000-05-24 Secr Defence Microprojectile delivery system
GB2365769A (en) 2000-08-18 2002-02-27 Secr Defence Skin preparations containing silicon
GB0104383D0 (en) 2001-02-22 2001-04-11 Psimedica Ltd Cancer Treatment
US6973718B2 (en) 2001-05-30 2005-12-13 Microchips, Inc. Methods for conformal coating and sealing microchip reservoir devices
CA2496736C (fr) 2002-08-23 2012-11-13 Mcmaster University Procedes et composes destines a reguler la morphologie et le retrecissement de silice derivee de silanes a polyol modifie
PL223153B1 (pl) 2002-09-18 2016-10-31 Allergan Inc Urządzenie do wszczepiania implantu okulistycznego
WO2004071949A2 (fr) 2003-02-13 2004-08-26 The Regents Of The University Of California Moulage nanostructure de polymeres organiques et de biopolymeres dans des matrices de silicium poreux
US7433811B2 (en) 2003-05-06 2008-10-07 The Regents Of The University Of California Direct patterning of silicon by photoelectrochemical etching
US7563451B2 (en) 2003-07-22 2009-07-21 Iowa State University Research Foundation, Inc. Capped mesoporous silicates
WO2005042023A1 (fr) 2003-10-21 2005-05-12 Psimedica Limited Materiau composite comprenant un semi-conducteur poreux impregne avec au moins une substance organique
GB0324483D0 (en) 2003-10-21 2003-11-19 Psi Medica Ltd Composite material
US20050181049A1 (en) 2003-11-19 2005-08-18 Dong Liang C. Composition and method for enhancing bioavailability
US7448867B2 (en) 2004-09-17 2008-11-11 Ormco Corporation Medical treatment apparatus
US20060067979A1 (en) * 2004-09-30 2006-03-30 Bausch & Lomb Incorporated Ophthalmic drug release device for multiple drug release
WO2006037160A1 (fr) 2004-10-05 2006-04-13 The University Of Melbourne Materiaux de polyelectrolyte poreux
US20090208556A1 (en) 2004-10-29 2009-08-20 Regents Of The University Of California, The Porous photonic crystals for drug delivery to the eye
WO2006067979A1 (fr) 2004-12-21 2006-06-29 Nec Corporation Boitier pour dispositif electrique a enveloppe par film
JP2007091716A (ja) * 2005-08-29 2007-04-12 Kuraray Co Ltd 水中薬剤徐放性微粒子及びその製造方法
GB0526332D0 (en) 2005-12-23 2006-02-01 Psimedica Ltd Pharmaceutical product
US8119394B2 (en) 2006-03-14 2012-02-21 University Of Rochester Cell culture devices having ultrathin porous membrane and uses thereof
BRPI0709672B8 (pt) * 2006-03-31 2021-06-22 3088922 Inc implante ocular insersível em um lúmen ocular e método de distribuição de um agente terapêutico a um olho
US20080057102A1 (en) 2006-08-21 2008-03-06 Wouter Roorda Methods of manufacturing medical devices for controlled drug release
CN101541836B (zh) 2006-09-25 2015-04-08 阿彻-丹尼尔斯-米德兰德公司 超吸收性的经表面处理的羧烷基化多糖及其制备方法
US20130084243A1 (en) 2010-01-27 2013-04-04 Liliane Goetsch Igf-1r specific antibodies useful in the detection and diagnosis of cellular proliferative disorders
CA2735899A1 (fr) 2008-09-25 2010-04-01 Cephalon, Inc. Formulations liquides de bendamustine
GB0817936D0 (en) 2008-09-30 2008-11-05 Intrinsiq Materials Global Ltd Porous materials
GB0817938D0 (en) 2008-09-30 2008-11-05 Intrinsiq Materials Global Ltd Cosmetic formulations
WO2010090596A1 (fr) 2009-02-04 2010-08-12 Agency For Science, Technology And Research Particule creuse de silice avec un polymère sur celle-ci
US20110300222A1 (en) 2009-02-20 2011-12-08 The Regents Of The University Of California Luminescent porous silicon nanoparticles, methods of making and using same
WO2010111627A1 (fr) 2009-03-26 2010-09-30 Psivida Us, Inc. Formulations implantables d'acides bisphosphoniques
JP5382700B2 (ja) 2009-03-27 2014-01-08 独立行政法人産業技術総合研究所 ロッド状多孔質シリカ粒子
JP5858906B2 (ja) 2009-05-04 2016-02-10 シヴィダ・ユーエス・インコーポレイテッドPsivida Us, Inc. 多孔質シリコン薬物溶出粒子
US20110028412A1 (en) 2009-08-03 2011-02-03 Cappellos, Inc. Herbal enhanced analgesic formulations
DE102010008982A1 (de) 2010-02-24 2011-08-25 Bayer Innovation GmbH, 40225 Siliciumhaltiges, biologisch degradierbares Material zur anti-inflammatorischen Therapie
GB201017048D0 (en) 2010-10-08 2010-11-24 Ucl Business Plc Composition
JP6026424B2 (ja) 2010-11-01 2016-11-16 シヴィダ・ユーエス・インコーポレイテッドPsivida Us, Inc. 治療薬送達のための生物浸食性ケイ素ベースのデバイス
WO2012088306A2 (fr) 2010-12-22 2012-06-28 Psivida Us, Inc. Dispositif d'administration de médicament injectable en deux pièces à élément d'obturation thermodurci
RU2013138180A (ru) 2011-02-11 2015-03-20 Псивида Юэс, Инк. Способы лечения макулярного отека с использованием противоотечных лекарственных средств
EP2691115A1 (fr) 2011-03-31 2014-02-05 Galderma Research & Development Compositions comprenant un produit de charge et au moins un matériau à base de silice biorésorbable et biodégradable
JP6100782B2 (ja) 2011-08-29 2017-03-22 キューエルティー インコーポレイテッド 緑内障および高眼圧症を治療するための活性剤の徐放送達
WO2014165108A1 (fr) 2013-03-12 2014-10-09 Psivida Us, Inc. Véhicules d'administration bioérodables à base de silicium pour l'administration d'agents thérapeutiques

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009009563A2 (fr) 2007-07-10 2009-01-15 The Regents Of The University Of California Matériaux et méthodes d'administration de compositions dans des tissus sélectionnés

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
"An Introduction to Microelectromechanical Systems Engineering", 2000, ARTECH HOUSE INC., pages: 265
"Design of Prodrugs", 1985, ELSEVIER SCIENTIFIC PUBLISHING CO.
"VLSI Technology", 1988, MCGRAW HILL, pages: 676
HALIMAOUI: "Properties of Porous Silicon", 1997, IEE, pages: 405
See also references of EP2427179A4

Cited By (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9566235B2 (en) 1998-04-17 2017-02-14 Psimedica Limited Implants for administering substances and methods of producing implants
US9962396B2 (en) 2009-05-04 2018-05-08 Psivida Us, Inc. Porous silicon drug-eluting particles
US9486459B2 (en) 2009-05-04 2016-11-08 Psivida Us, Inc. Porous silicon drug-eluting particles
US9333173B2 (en) 2010-11-01 2016-05-10 Psivida Us, Inc. Bioerodible silicon-based devices for delivery of therapeutic agents
JP2016121173A (ja) * 2010-11-01 2016-07-07 シヴィダ・ユーエス・インコーポレイテッドPsivida Us, Inc. 治療薬送達のための生物浸食性ケイ素ベースのデバイス
EP2635255A4 (fr) * 2010-11-01 2016-11-09 Psivida Inc Dispositifs à base de silicium pouvant être biologiquement érodés pour administration d'agents thérapeutiques
US9808421B2 (en) 2010-11-01 2017-11-07 Psivida Us, Inc. Bioerodible silicon-based devices for delivery of therapeutic agents
WO2012061377A1 (fr) * 2010-11-01 2012-05-10 Psivida Us, Inc. Dispositifs à base de silicium pouvant être biologiquement érodés pour administration d'agents thérapeutiques
AU2016259330B2 (en) * 2010-11-01 2018-11-08 Eyepoint Pharmaceuticals Us, Inc. Bioerodible silicon-based devices for delivery of therapeutic agents
US11026885B2 (en) 2010-11-01 2021-06-08 Eyepoint Pharmaceuticas, Inc. Bioerodible silicon-based devices for delivery of therapeutic agents
JP2014508749A (ja) * 2011-02-11 2014-04-10 サイヴィーダ ユーエス,インコーポレイテッド 抗浮腫治療薬を使用する黄斑浮腫の治療方法
US10835495B2 (en) 2012-11-14 2020-11-17 W. R. Grace & Co.-Conn. Compositions containing a biologically active material and a non-ordered inorganic oxide material and methods of making and using the same
US9603801B2 (en) 2013-03-15 2017-03-28 Psivida Us, Inc. Bioerodible silicon-based compositions for delivery of therapeutic agents
US9980911B2 (en) 2013-03-15 2018-05-29 Psivida Us, Inc. Bioerodible silicon-based compositions for delivery of therapeutic agents

Also Published As

Publication number Publication date
AU2010246067B2 (en) 2016-07-07
US9962396B2 (en) 2018-05-08
JP5858906B2 (ja) 2016-02-10
US20150272965A1 (en) 2015-10-01
CN105476951A (zh) 2016-04-13
US9486459B2 (en) 2016-11-08
CN102405042A (zh) 2012-04-04
US20100278931A1 (en) 2010-11-04
WO2010129545A3 (fr) 2011-03-24
JP2012526048A (ja) 2012-10-25
CA2758607A1 (fr) 2010-11-11
AU2016203393B2 (en) 2017-07-27
US20170128464A1 (en) 2017-05-11
JP2015143257A (ja) 2015-08-06
EP2427179A2 (fr) 2012-03-14
AU2010246067A1 (en) 2011-11-10
CA2758607C (fr) 2018-05-15
US9023896B2 (en) 2015-05-05
JP6085634B2 (ja) 2017-02-22
EP2427179A4 (fr) 2013-09-11
CN103494765A (zh) 2014-01-08
AU2016203393A1 (en) 2016-06-16

Similar Documents

Publication Publication Date Title
US9962396B2 (en) Porous silicon drug-eluting particles
US9849085B2 (en) Sustained release drug delivery devices, methods of use, and methods of manufacturing thereof
EP1347741B1 (fr) Dispositifs d'administration de medicament a liberation prolongee pour la diffusion d'agents multiples
CA2432225C (fr) Dispositifs d'administration de medicament a liberation prolongee a tampons permeables prefabriques
US6756049B2 (en) Sustained release drug delivery devices
US5902598A (en) Sustained release drug delivery devices
US20060034929A1 (en) Sustained release drug delivery devices with prefabricated permeable plugs
US20040062787A1 (en) Therapeutic combination of amlodipineand benazepril/benazeprilat
EP1847255A2 (fr) Dispositifs de livraison de médicaments à libération prolongée avec noyaux de médicaments recouverts

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201080017208.4

Country of ref document: CN

121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 10772695

Country of ref document: EP

Kind code of ref document: A2

WWE Wipo information: entry into national phase

Ref document number: 2010772695

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2758607

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2012508826

Country of ref document: JP

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2010246067

Country of ref document: AU

Date of ref document: 20100504

Kind code of ref document: A